You are on page 1of 11

This article was downloaded by: [University of Arizona]

On: 22 January 2013, At: 04:49


Publisher: Routledge
Informa Ltd Registered in England and Wales Registered Number: 1072954 Registered office: Mortimer House,
37-41 Mortimer Street, London W1T 3JH, UK

Nutrition and Cancer


Publication details, including instructions for authors and subscription information:
http://www.tandfonline.com/loi/hnuc20

Inhibition of IGF-1 Signaling by Genistein: Modulation


of E-Cadherin Expression and Downregulation of β-
Catenin Signaling in Hormone Refractory PC-3 Prostate
Cancer Cells
a b c d a
Joomin Lee , Jihyeung Ju , Seyeon Park , Sung Joon Hong & Sun Yoon
a
Department of Food and Nutrition, Brain Korea 21 Project, Yonsei University College of
Human Ecology, Seodaemun-Gu, Seoul, Korea
b
Department of Food and Nutrition, Chungbuk National University, Chungbuk, Korea
c
Department of Applied Chemistry, Dongduk Women's University, Seoul, Korea
d
Department of Urology, Urological Science Institute, Brain Korea 21 Project for Medical
Sciences, Yonsei University College of Medicine, Seoul, Korea
Version of record first published: 18 Nov 2011.

To cite this article: Joomin Lee , Jihyeung Ju , Seyeon Park , Sung Joon Hong & Sun Yoon (2012): Inhibition of IGF-1 Signaling
by Genistein: Modulation of E-Cadherin Expression and Downregulation of β-Catenin Signaling in Hormone Refractory PC-3
Prostate Cancer Cells, Nutrition and Cancer, 64:1, 153-162

To link to this article: http://dx.doi.org/10.1080/01635581.2012.630161

PLEASE SCROLL DOWN FOR ARTICLE

Full terms and conditions of use: http://www.tandfonline.com/page/terms-and-conditions

This article may be used for research, teaching, and private study purposes. Any substantial or systematic
reproduction, redistribution, reselling, loan, sub-licensing, systematic supply, or distribution in any form to
anyone is expressly forbidden.

The publisher does not give any warranty express or implied or make any representation that the contents
will be complete or accurate or up to date. The accuracy of any instructions, formulae, and drug doses should
be independently verified with primary sources. The publisher shall not be liable for any loss, actions, claims,
proceedings, demand, or costs or damages whatsoever or howsoever caused arising directly or indirectly in
connection with or arising out of the use of this material.
Nutrition and Cancer, 64(1), 153–162
Copyright C 2012, Taylor & Francis Group, LLC

ISSN: 0163-5581 print / 1532-7914 online


DOI: 10.1080/01635581.2012.630161

Inhibition of IGF-1 Signaling by Genistein: Modulation of


E-Cadherin Expression and Downregulation of ␤-Catenin
Signaling in Hormone Refractory PC-3 Prostate Cancer Cells
Joomin Lee
Department of Food and Nutrition, Brain Korea 21 Project, Yonsei University College of Human
Ecology, Seodaemun-Gu, Seoul, Korea

Jihyeung Ju
Department of Food and Nutrition, Chungbuk National University, Chungbuk, Korea

Seyeon Park
Downloaded by [University of Arizona] at 04:49 22 January 2013

Department of Applied Chemistry, Dongduk Women’s University, Seoul, Korea


Sung Joon Hong
Department of Urology, Urological Science Institute, Brain Korea 21 Project for Medical Sciences,
Yonsei University College of Medicine, Seoul, Korea

Sun Yoon
Department of Food and Nutrition, Brain Korea 21 Project, Yonsei University College of Human
Ecology, Seodaemun-Gu, Seoul, Korea

inhibited cell growth in IGF-1-stimulated PC-3 cells, possibly by


Elevated levels of insulin-like growth factor-1 (IGF-1) are asso- inhibiting downstream of IGF-1R activation.
ciated with an increased risk of several different cancers, including
prostate cancer. Inhibition of IGF-1 and the downstream signal-
ing pathways mediated by the activation of the IGF-1 receptor
(IGF-1R) may be involved in inhibiting prostate carcinogenesis. We
investigated whether genistein downregulated the IGF-1/IGF-1R
INTRODUCTION
signaling pathway and inhibited cell growth in hormone refractory Prostate cancer is a common malignancy that is the sec-
PC-3 prostate cancer cells. Genistein treatment caused a significant ond leading cause of cancer-related deaths in Western males
inhibition of IGF-1-stimulated cell growth. Flow cytometry anal- (1). Most early prostate cancers require androgen stimulation
ysis revealed that genistein significantly decreased the number of (androgen-dependent), and blocking androgen action through
IGF-1-stimulated cells in the G0/G1 phase of the cell cycle. In
IGF-1-treated cells, genistein effectively inhibited the phosphory- androgen ablation is a common therapy for disease. However,
lation of IGF-1R and the phosphorylation of its downstream tar- after a time, prostate cancer progresses toward a hormone re-
gets, such as Src, Akt, and glycogen synthase kinase-3β (GSk-3β). fractory (androgen-independent) state and there are no effective
IGF-1 treatment decreased the levels of E-cadherin but increased therapeutic options for this stage of disease (2,3). Deregulation
the levels of β-catenin and cyclin D1. However, genistein treat- of specific growth factors is associated with the development of
ment greatly attenuated IGF-1-induced β-catenin signaling that
correlated with increasing the levels of E-cadherin and decreasing androgen ablation-refractory prostate cancer. Laboratory- and
cyclin D1 levels in PC-3 cells. In addition, genistein inhibited T- population-based studies have linked elevated levels of insulin-
cell factor/lymphoid enhancer factor (TCF/LEF)-dependent tran- like growth factor-1 (IGF-1) to prostate cancer progression (4).
scriptional activity. These results showed that genistein effectively IGF-1 is postulated to contribute to prostate cancer development
by blocking apoptosis, which promotes cancer cell prolifera-
tion and invasion. The actions of IGF-1 are mediated by the
Submitted 9 May 2010; accepted in final form 25 August 2011. insulin-like growth factor-1 receptor (IGF-1R). IGF-1 binding
Address correspondence to Sun Yoon, Department of Food and
to IGF-1R allows the β-subunits of IGF-1R to display intrinsic
Nutrition, Yonsei University 262 Seongsanno, Seodaemun-gu, Seoul
120-749, Korea. Phone: +82 2 2123 3119. Fax: +82 2 365 3118. tyrosine kinase activity and activates downstream signals via
E-mail: snkim@yonsei.ac.kr phosphorylation of key proteins.

153
154 J. LEE ET AL.

IGF-1R also affects the expression or phosphorylation of dif- CA). Genistein was dissolved in DMSO (final concentration
ferent cell–cell adhesion proteins (5). The E-cadherin/β-catenin <0.05%). β-catenin, E-cadherin, IGF-1Rβ, phospho-IGF-1Rβ
complex plays an important role in maintaining proper epithelial (Tyr 1135/1136), Src, phospho-Src (Tyr 416), Akt, phospho-
cell function and tissue integrity (6). IGF-1 treatment decreases Akt (Ser 473), GSK-3β, phospho-GSK-3β (Ser 9), cyclin D1,
E-cadherin expression and reduces cell–cell adhesion (7). Loss β-actin, and histone H2A antibodies, as well as antirabbit and
of E-cadherin expression commonly occurs when normal cells antimouse peroxidase-conjugated antibodies, were purchased
transit to highly malignant human epithelial cancers, such from Cellular Signaling Technology (Beverly, MA). Total-ERK
as prostate cancer (8,9). and phospho-ERK (Thr202/Tyr204) antibodies were purchased
IGF-1 induction enhances the tyrosine phosphorylation of from Santa Cruz Biotechnology (Santa Cruz, CA).
β-catenin. This results in the dissociation of β-catenin from
the E-cadherin complex and increase in the cytoplasmic level Cell Viability Assay
of β-catenin (10). β-catenin is not only involved structurally PC-3 cells were seeded into 96-well plates at 5 × 103
in the adherent junction complex but also acts as a signaling cells/well. The following day, the 10% FBS medium was re-
molecule in the Wnt signaling pathway (11). The cytoplasmic moved, and the cells were washed once with phosphate-buffered
expression of β-catenin is low in normal cells because it is saline (PBS). Fresh serum-free medium with or without genis-
easily targeted for ubiquitination by glycogen synthase kinase- tein and IGF-1 (50 ng/mL) was provided. Cell viability was
3β (GSK-3β) phosphorylation and is degraded by proteosomes determined after 24 and 48 h using the CellTiter96 Aqueous-
Downloaded by [University of Arizona] at 04:49 22 January 2013

(12,13). In cancer cells, β-catenin accumulates in the cytoplasm One-Solution Assay (Promega, Madison, WI), according to the
and translocates into the nucleus, where it interacts with T-cell manufacturer’s protocol.
factor/lymphoid enhancer factor (TCF/LEF) factors to activate
Cell Cycle Analysis by Flow Cytometry
specific target genes, such as c-myc and cyclin D1, to promote
Cells (5 × 105) were plated into 10-cm-diameter dishes and
cancer progression.
allowed to adhere overnight. Cells were serum starved for 24 h
IGF-1/IGF-1R plays a role in maintaining the malignant phe-
and then treated with different concentrations of genistein and
notype and enhanced activity of IGF-1R has been observed in
IGF-1 for an additional 24 h. Treated cells were harvested for cell
prostate cancers (14). An increase in IGF-1R expression is ob-
cycle analysis. Cells were washed in PBS, fixed in 70% ethanol,
served in the majority of metastatic and androgen-independent
and stored at −20◦ C. Before analysis, cells were washed once
prostate cancer specimens as compared to primary disease
with PBS and PI was added. Samples were incubated at room
(15–17). IGF-1/IGF-1R pathways are related to activation of
temperature in the dark for 15 min. DNA content/cell number
cellular growth and proliferation signals via protein tyrosine
(10,000 cells per sample) was analyzed by flow cytometry using
phosphorylation. Genistein, a soy-derived isoflavone, has re-
a FACSCalibur apparatus (Becton Dickinson, San Jose, CA).
ceived much attention as a dietary component that acts as a
The percentage of cells in each phase of the cell cycle was
tyrosine kinase inhibitor to block or reverse carcinogenesis, in-
determined by the Modfit LT program (Verity Software House,
cluding prostate cancers (18). The present study investigated
Topsham, ME).
whether genistein could overcome the growth stimulatory ef-
fects of IGF-1 in hormone refractory prostate cancer cells by in- Preparation of Cell Extracts
hibiting the activity of specific receptor tyrosine kinase (RTKs) Following genistein and IGF-1 treatment, the medium was
and related downstream pathways of signal transduction, in- aspirated and the cells were washed with cold PBS. Total cell
cluding the Wnt signaling pathway. extracts were prepared by scraping the cells into lysis buffer [1%
NP-40, 0.5% sodium deoxycholate, 0.1% sodium dodecyl sul-
MATERIALS AND METHODS fate (SDS), 10 µl/ml phenylmethanesulfonyl fluoride (PMSF),
30 µl/ml aprotinin, and 10 µl/ml sodium orthovanadate]. Nu-
Cell Culture clear and cytoplasmic proteins were isolated using the CelLytic
The androgen insensitive, p53-negative PC-3 human prostate NuCLEAR Extraction kit (Sigma, St. Louis, MO). Extracts were
cancer cells were obtained from the Korean Cell Line aliqouted and stored at −80◦ C for subsequent Western blot anal-
Bank (Korea). The cell line was grown in RPMI 1640 yses. Protein concentrations were determined by the Bradford
(Gibco/Invitron, Carlsbad, CA) medium supplemented with Protein Assay (Bio-Rad, Hercules, CA).
10% heat-inactivated fetal bovine serum (FBS; Gibco), 100
units/ml penicillin, and 100 µg/ml streptomycin in an incubator SDS-Polyacrylamide Gel Electrophoresis
at 37◦ C with 5% CO2 . and Western Blotting
Cell extracts (15–50 µg of protein) were electrophoresed
Chemicals and Antibodies on SDS-polyacrylamide gels and transferred to nitrocellulose
Genistein, IGF-1, and dimethyl sulfoxide (DMSO) were membranes. Blots were blocked in 5% nonfat dry milk/TBS
purchased from Sigma (St. Louis, MO). Propidium io- containing 0.05% Tween-20 and incubated overnight at 4◦ C
dide (PI) was purchased from BD Pharmingen (San Diego, with primary antibody. The membranes were then hybridized
GENISTEIN MODULATES β-CATENIN IN PROSTATE CANCER CELLS 155

with the secondary antibody conjugated to HRP for 1 h at room the ratio of cells in S phase. There was a decrease in the per-
temperature. Immuno reactions were detected using ECL Plus centage of cells in G0/G1 and an increase in the percentage
Western blotting reagents (Amersham Pharmacia Biotech, Baie of cells in G2/M following treatment with genistein and IGF-1
d’Urfe, Québec, Canada). Quantitative analysis was performed (Fig. 1C).
by volume densitometry after scanning the film.

Immunofluorescence Staining and Confocal Genistein Inhibits IGF-1-Induced Tyrosine


Laser Microscopy Phosphorylation of IGF-1R and Src
Cells (1.5 × 105/well) were seeded onto glass coverslips, IGF-1R is a protein tyrosine kinase receptor that is involved
grown for 24 h to obtain compact cell monolayers, and then in the proliferation and differentiation of normal cells. IGF-
fixed in 3.7% formaldehyde for 15 min. Cells were saturated in 1/IGF-1R plays a key role in the growth promoting-function
PBS/1% bovine serum albumin (PBS/BSA) with 0.1% Triton X- of prostate cancer cells (19,20). To determine whether genis-
100 and incubated with primary antibodies diluted in PBS/BSA tein inhibited IGF-1-induced phosphorylation of IGF-1R, PC-3
for 60 min at room temperature. After 3 washes in PBS, sec- cells were treated with IGF-1 (50 ng/mL) in the absence or
ondary fluorochrome-conjugated (Alexa Fluor 488 and Alexa presence of genistein. A 30-min treatment with IGF-1 alone
Fluor 546) antibodies in PBS/BSA were added for 60 min. Cov- showed the highest level of IGF-1R phosphorylation (Fig. 2A).
erslips were mounted on glass slides with mounting solution and However, phosphorylation of IGF-1R decreased significantly
when the cells were cotreated with 25 or 50 µM genistein in the
Downloaded by [University of Arizona] at 04:49 22 January 2013

analyzed by confocal microscopy (LSM 510 Confocal Micro-


scope, Carl Zeiss, Inc., Thornwood, NY). Nuclei were stained presence of IGF-1 (50 ng/mL) for 30 min (Fig. 2B). We also
with 4 ,6-diamidino-2-phenylindole (DAPI) for 5 min, and the evaluated the effect of genistein on phosphorylation of the Src
coverslips were mounted onto glass slides with mounting solu- nonreceptor protein tyrosine kinase. Src is one of the most im-
tion. Slides were analyzed by confocal microscopy (LSM 510 portant intracellular oncogenic signals and it participates in the
Confocal Microscope, Carl Zeiss, Inc.). androgen-independent growth of prostate cancer cells (21–23).
IGF-1 treatment induced Src phosphorylation (Fig. 2C). A 30-
Transfection and Luciferase Reporter Gene Assay min stimulation with genistein significantly abrogated the IGF-
Cells were transfected with reporter gene constructs using 1-induced upregulation of Src phosphorylation. Total Src pro-
Lipofectamine 2000 (Invitrogen, Carlsbad, CA) according to tein levels were not affected in treated cells (Fig. 2D).
the manufacturer’s instructions. Firefly and Renilla luciferase
activities were sequentially determined 24 h posttransfection Genistein Suppresses IGF-1-Induced Phosphorylation
using the Dual Glo luciferase assay system (Promega, Madison, of PI3K/Akt and GSK-3␤
WI) according to the manufacturer’s instructions. IGF-1R signaling involves PI3K/Akt phosphorylation that
can promote prostate cancer cell growth (24). To investigate
Statistical Analyses
whether genistein suppressed the PI3K/Akt pathway, the phos-
All the results were presented as mean values ± SD. Data
phorylation levels of Akt and GSK-3β were assessed. IGF-1
from each assay were statistically analyzed by 1-way ANOVA
treatment caused an increase in Akt phosphorylation that was
with Bonferroni’s test. Differences were considered significant
the most prominent at 30 min following treatment (Fig. 3A).
when the P value was less than 0.05.
However, Akt phosphorylation was significantly inhibited by
genistein in a dose-dependent manner when compared with
RESULTS IGF-1 treatment alone (Fig. 3B). PC-3 cells treated for 24 h with
IGF-1 had the highest level of GSK-3β phosphorylation (Fig.
Genistein Decreases IGF-1-Stimulated PC-3 3C). This effect was inhibited by genistein in a dose-dependent
Cell Proliferation manner (Fig. 3D).
We examined the effect of genistein on PC-3 cell prolifera-
tion in the presence or absence of IGF-1 stimulation using the
MTS assay. IGF-1 significantly increased PC-3 cell prolifera- Genistein Does Not Alter IGF-1-Induced
tion when added to serum-free medium (Fig. 1A). Genistein Phosphorylation of ERK1/2
inhibited the IGF-1 (50 ng/mL) stimulated PC-3 cell growth Because the ras-raf-MAPK pathway is the important down-
in a dose- and time-dependent manner as compared to IGF-1 stream of IGF-1R, we examined the levels of phosphorylated
treatment alone (Fig. 1B). Genistein had no inhibitory effect on and total ERK1/2 by Western blots. As shown in Fig. 4A, the
PC-3 cell growth in the absence of IGF-1. The distribution of ratio of phosphorylated to total ERK1/2 protein was highest fol-
PC-3 cells in the cell cycle was analyzed to further investigate lowing IGF-1 treatment for 30 min. Treatment with genistein
the mechanism of genistein-induced suppression of cell growth. (1–50 µM), with or without IGF-1, did not significantly affect
Approximately 75% of cells were blocked in G0/G1 following a the ratio of phosphorylated to total ERK1/2 protein at 30 min
24-h serum starvation. IGF-1 stimulation significantly increased (Fig. 4B).
156 J. LEE ET AL.
Downloaded by [University of Arizona] at 04:49 22 January 2013

FIG. 1. Genistein inhibited insulin-like growth factor-1 (IGF-1)-induced cell proliferation in PC-3 cells. A: Cells were starved in serum-free media for 24 h and
then treated with different concentrations of IGF-1 for an additional 24 h. Number of viable cells (% of control) was measured by the MTS assay. B: Cells were
starved in serum-free media for 24 h and then treated with different concentrations of genistein for 24 h or 48 h in the absence or presence of IGF-1 (50 ng/mL).
Number of viable cells (% of control) was measured by the MTS assay. C: Cells were treated with genistein at different concentrations for 24 h in the presence of
IGF-1 (50 ng/mL). The cell cycle distribution was analyzed by flow cytometry. Data are shown as mean ± SD of triplicate determinations (n = 3). Significantly
different (P < 0.05) compared to treatment with dimethyl sulfoxide (a) and treatment with IGF-1 alone (b) by 1-way ANOVA followed by Bonferroni’s test.

Genistein Alters E-Cadherin and ␤-Catenin Levels 25, or 50 µM of genistein for 24 h increased E-cadherin protein
IGF-1R mediated signaling can result in decreased intracel- levels by 104.1%, 119.4%, 162.7%, and 242.4%, respectively
lular adhesion due to increased tyrosine phosphorylation of E- (Fig. 5A) as compared to IGF-1 treatment alone. Genistein and
cadherin and β-catenin (25). Many studies have shown that the IGF-1 treatment also decreased the nuclear and cytoplasmic
alterations in E-cadherin/β-catenin expression are linked to dis- levels of β-catenin. Reductions of 92.3%, 74.6%, 68.5%, and
ease progression via a loss of cell–cell adhesion and increased 33.2% in nuclear β-catenin levels and 77.4%, 60%, 47.4%,
metastasis (26–28). Therefore, we investigated the effects of and 40.3% in cytoplasmic levels were observed with 1, 10, 25,
genistein and IGF-1 cotreatment on E-cadherin and β-catenin and 50 µM of genistein and IGF-1 cotreatment for 24 h, respec-
in PC-3 cells. Cotreatment of PC-3 cells with IGF-1 and 1, 10, tively (Fig. 5B). Fluorescence microscopy also revealed altered
GENISTEIN MODULATES β-CATENIN IN PROSTATE CANCER CELLS 157
Downloaded by [University of Arizona] at 04:49 22 January 2013

FIG. 2. Genistein decreased the tyrosine phosphorylation of insulin-like growth factor-1 receptor (IGF-1R) and Src in insulin-like growth factor-1 (IGF-1)-treated
PC-3 cells. A, C: Cells were serum-starved for 24 h and then treated with 50 ng/mL IGF-1 for different times. The total and phosphorylated levels of IGF-1R
and Src were measured by Western blot analysis. Results are shown as mean fold change of 2 measurements. B, D: Cells were treated with genistein at different
concentrations for 30 min in the presence of IGF-1 (50 ng/mL). The total and phosphorylated levels of IGF-1R and Src were measured by Western blot analysis.
Results are shown as mean ± SD of 3 independent experiments (n = 3). Significantly different (P < 0.05) compared to treatment with dimethyl sulfoxide (a) and
treatment with IGF-1 alone (b) by 1-way ANOVA followed by Bonferroni’s test.

E-cadherin and β-catenin levels after IGF-1 and/or genistein with the TOPFlash TCF/LEF luciferase reporter construct.
treatment (50 µM) (Fig. 5C). Luciferase activity was significantly decreased in PC-3 prostate
cancer cells following treatment with 25 and 50 µM genistein
(Fig. 6A). Since β-catenin signaling can induce or repress the
Genistein Treatment Decreases ␤-Catenin-Mediated expression of a variety of target genes, we assessed the protein
Gene Transcription expression of cyclin D1, a transcriptional target of β-catenin,
To determine if genistein treatment could reduce β-catenin- by Western blot. Treatment of PC-3 cells with 50 µM genistein
mediated gene transcription, cells were transiently transfected for 24 h decreased the nuclear level of cyclin D1 by 25.5% and
158 J. LEE ET AL.
Downloaded by [University of Arizona] at 04:49 22 January 2013

FIG. 3. Genistein decreased the phosphorylation of Akt and GSK-3β in insulin-like growth factor-1 (IGF-1)-treated PC-3 cells. A, C: Cells were serum-starved
for 24 h and then treated with 50 ng/mL IGF-1 for different times. Total and phosphorylated levels of Akt and glycogen synthase kinase-3β (GSK-3β) were
measured by Western blot analysis. Results are shown as mean fold change of 2 measurements. B: Cells were treated for 30 min with genistein at different
concentrations in the presence of IGF-1 (50 ng/mL). Total and phosphorylated levels of Akt were measured by Western blot analysis. D: Cells were treated for
24 h with genistein at different concentrations in the presence of IGF-1 (50 ng/mL). Total and phosphorylated levels of GSK-3β were measured by Western blot
analysis. Results are shown as mean ± SD of 3 independent experiments (n = 3) (B, D). Significantly different (P < 0.05) compared to treatment with dimethyl
sulfoxide (a) and treatment with IGF-1 alone (b) by 1-way ANOVA followed by Bonferroni’s test.

the cytoplasmic level by 60.3% as compared to IGF-1 alone inhibited by genistein. IGF-1 stimulation increased the phos-
(Fig. 6B). phorylated form of the IGF-1R protein and its downstream
signaling protein Src. The Src family of kinases (SFK) are
DISCUSSION highly expressed in androgen-independent prostate cancer cell
We showed that IGF-1 increased proliferation of PC-3 lines and are responsible for multiple signal transduction events,
prostate cancer cells, and this increased effect was significantly such as differentiation, adhesion, and migration (29). Our results
GENISTEIN MODULATES β-CATENIN IN PROSTATE CANCER CELLS 159
Downloaded by [University of Arizona] at 04:49 22 January 2013

FIG. 4. Genistein did not significantly decrease ERK 1/2 phosphorylation. A: Cells were serum-starved for 24 h and then treated with 50 ng/mL insulin-like
growth factor-1 (IGF-1) for different times. Results are shown as mean fold change of 2 measurements. B: Cells were treated with different concentrations of
genistein for 30 min in the presence of IGF-1 (50 ng/mL). Total and phosphorylated levels of ERK 1/2 were measured by Western blot analysis. Results are shown
as mean ± SD of 3 independent experiments (n = 3). Significantly different (P < 0.05) compared to treatment with dimethyl sulfoxide (a) by 1-way ANOVA
followed by Bonferroni’s test.

showed that treatment of PC-3 cells with genistein inhibited the naling pathway through inhibition of IGF-1-initiated phospho-
phosphorylation of IGF-1R and Src, which were activated with rylation of specific RTKs.
IGF-1 stimulation. The Wnt signaling pathway has recently emerged as an im-
Several studies have shown that a number of RTKs and the ty- portant player in prostate tumorigenesis. Only a small percent-
rosine kinase Src induce tyrosine phosphorylation of β-catenin. age of prostate cancer samples harbor mutations in the destruc-
This releases β-catenin from the cadherin–catenin complexes tion complex and β-catenin itself, suggesting that other mecha-
and allows it to participate in the Wnt signaling pathway (30). nisms may be involved in activating the Wnt signaling pathway
Both E-cadherin and β-catenin co-localize at the surface of in prostate cancer progression. β-catenin, a key component of
epithelial cells and play crucial roles in epithelial cell–cell ad- Wnt signaling, has multifunctional roles through its associations
hesion (31). Disruption of the cadherin–catenin complex has with various protein partners. β-catenin is involved in cell–cell
been linked to the invasion and metastasis of various cancer adhesion when associated with cadherins, and it regulates gene
cells, including prostate cancer (32–35). transcription when associated with the TCF/LEF proteins. Sev-
We demonstrated that treatment of PC-3 cells with genistein eral studies have shown that increased nuclear localization of
and IGF-1 significantly increased E-cadherin expression and β-catenin and its transcriptional activity are important events
decreased β-catenin accumulation, as compared to treatment during cancer development (33).
with IGF-1 alone. Genistein-induced increases in E-cadherin We demonstrated that IGF-1 increased nuclear β-catenin lev-
expression and decreases in phospho-Src levels may contribute els and the subsequent activation of TCF/LEF transcription fac-
to the restoration of the E-cadherin/β-catenin complex. Further tor family members. IGF-1 also increased cyclin D1, one of the
studies are necessary to fully demonstrate such an association. target genes of the β-catenin/TCF complex, and promoted the
Since IGF-1 is one of the most potent natural activators S cell cycle transition leading to cancer cell proliferation. How-
of the Akt signaling pathway, we investigated whether genis- ever, genistein blocked IGF-1-mediated downstream signals by
tein altered the phosphorylation status of Akt/GSK-3β in PC-3 dephosphorylating IGF-1R, Src, Akt, and GSK-3β and altered
cells stimulated with IGF-1. IGF-1 increased the phosphoryla- E-cadherin and β-catenin levels in PC-3 prostate cancer cells.
tion of Akt/GSK-3β, and this effect was reversed by genistein. Treatment of cells with genistein and IGF-1 decreased the per-
Genistein-mediated dephosphorylation and activation of GSK- centage of cells in the G0/G1phases and increased the number
3β may enhance the phosphorylation of β-catenin, leading to of cells in the G2/M phases. Animal studies have shown that
its ubiquitylation and proteasome-mediated degradation. The genistein treatment decreased the expression of cyclin D1 and
present results suggest that genistein may inhibit the Wnt sig- reduced tumor growth (36).
160 J. LEE ET AL.
Downloaded by [University of Arizona] at 04:49 22 January 2013

FIG. 5. Genistein modulated the protein levels of E-cadherin and β-catenin in insulin-like growth factor-1 (IGF-1)-treated PC-3 cells. Cells were starved in
serum-free medium for 24 h and then treated with different concentrations of genistein and 50 ng/mL IGF-1 for 12 h and 24 h. A: Western blots analysis of
E-cadherin in whole cell lysates. E-cadherin levels were quantified by volume densitometry and normalized to β-actin. B: Western blot analysis of β-catenin in
cytoplasmic and nuclear fractions. Cytoplasmic and nuclear β-catenin levels (quantified by volume densitometry) were normalized to β-actin and histone H2A,
respectively. Western blot results are shown as mean ± SD of 3 independent experiments (n = 3). C: Confocal microscopic analysis of E-cadherin and β-catenin.
DAPI was used for staining the nucleus. The results are representative of 3 independent experiments that showed similar staining patterns. Significantly different
(P < 0.05) compared to treatment with dimethyl sulfoxide (a) and treatment with IGF-1 alone (b) by 1-way ANOVA followed by Bonferroni’s test.
GENISTEIN MODULATES β-CATENIN IN PROSTATE CANCER CELLS 161
Downloaded by [University of Arizona] at 04:49 22 January 2013

FIG. 6. Genistein inhibited β-catenin-mediated gene transcription and cyclin D1 expression in insulin-like growth factor-1 (IGF-1)-treated PC-3 cells. A:
Cells were transiently transfected with TOPflash and FOPflash reporter gene constructs. Twenty-four hours after transfection, cells were treated with different
concentrations of genistein and IGF-1 (50 ng/mL). B: Nontransfected cells were starved in serum-free medium for 24 h and then treated with 50 µM genistein
for an additional 24 h in the presence of 50 ng/mL IGF-1. Nuclear and cytosolic cyclin D1 levels were measured by Western blot analysis and normalized to
histone H2A and β-actin, respectively. Nuclear protein, as represented by histone H2A, was not detected in the cytoplasmic fractions. This indicates that there
was no significant cross-contamination between nuclear and cytoplasmic fractions in our preparation. Representative results of 3 independent experiments are
shown. Significantly different (P < 0.05) compared to treatment with dimethyl sulfoxide (a) and treatment with IGF-1 alone (b) by 1-way ANOVA followed by
Bonferroni’s test.

After androgen deprivation at late stage in prostate cancer, tein also increases E-cadherin and decreases β-catenin, which
tumor of castration-resistant prostate cancer (CRPC) continues results in inhibition of TCF/LEF transcription factor binding to
to depend on androgen receptor (AR). IGF-1/IGF-1R is one Wnt target gene promoters in PC-3 prostate cancer cells. These
of the key signaling pathways that activate AR (37). Our stud- observations indicate that genistein may play a role in the pre-
ies in PC-3 cells showed that, in absence of AR, IGF-1 al- vention and/or treatment of hormone refractory prostate cancer.
ternatively modulated E-cadherin and β-catenin pathways and
genistein decreased IGF-1/IGF-1R mediated downstream sig-
nals. Further studies are needed to consider AR and E-cadherin ACKNOWLEDGMENTS
/β-catenin pathways with genistein treatment. Genistein plasma
This study was supported by a grant of the Korea Health
levels in people on a soy-rich diet are in the 1–5 µM range af-
21 R&D project, Ministry of Health and Welfare, Republic of
ter metabolism and excretion (38,39). At low doses (<10 µM),
Korea (Contact grant number A050611).
genistein stimulated the growth of estrogen-sensitive cell lines
(40), while decreasing proliferation at higher doses (>10–20
µM) (41). This suggests that genistein exerts a biphasic effect
on growth and proliferation of cancer cells. Genistein has multi- REFERENCES
1. Debes JD and Tindall DJ: Mechanisms of androgen-refractory prostate
ple biochemical effects and can act as an antiproliferative agent
cancer. N Engl J Med 351, 1488–1490, 2004.
at high concentrations (≥10 µM). 2. Schröder FH: Progress in understanding androgen-independent prostate
In conclusion, genistein exerts its role as a critical inhibitor cancer (AIPC): a review of potential endocrine-mediated mechanisms. Eur
of RTKs through a series of signal transduction events. Genis- Urol 53, 1129–1137, 2008.
162 J. LEE ET AL.

3. Miyamoto H, Messing EM, and Chang C: Androgen deprivation therapy for Etk/Bmx, Src, and focal adhesion kinase. Mol Cell Biol 21, 8385–8397,
prostate cancer: current status and future prospects. Prostate 61, 332–353, 2001.
2004. 22. Lee LF, Louie MC, Desai SJ, Yang J, Chen HW, et al.: Interleukin-8 confers
4. Renehan AG, Zwahlen M, Minder C, O’Dwyer ST, Shalet SM, et al.: androgen-independent growth and migration of LNCaP: differential effects
Insulin-like growth factor (IGF)-I, IGF binding protein-3, and cancer risk: of tyrosine kinases Src and FAK. Oncogene 23, 2197–2205, 2004.
systematic review and meta-regression analysis. Lancet 363, 346–353, 23. Unni E, Sun S, Nan B, McPhaul MJ, Cheskis B, et al.: Changes in androgen
2004. receptor nongenotropic signaling correlate with transition of LNCaP cells
5. Mauro L, Salerno M, Morelli C, Boterberg T, Bracke ME, et al.: Role of to androgen independence. Cancer Res 64, 7156–7168, 2004.
the IGF-I receptor in the regulation of cell–cell adhesion: implications in 24. Grimberg A: Mechanisms by which IGF-I may promote cancer. Cancer
cancer development and progression. J Cell Physiol 194, 108–116, 2003. Biol Ther 2, 630–635, 2003.
6. Nelson WJ and Nusse R: Convergence of Wnt, beta-catenin, and cadherin 25. Bahr C and Groner B: The IGF-1 receptor and its contributions to metastatic
pathways. Science 5, 1483–1487, 2004. tumor growth-novel approaches to the inhibition of IGF-1R function.
7. Ozawa M and Kemler R: Altered cell adhesion activity by pervanadate due Growth Factors 23, 1–14, 2005.
to the dissociation of alpha-catenin from the E-cadherin.catenin complex. 26. Jaggi M, Johansson SL, Baker JJ, Smith LM, Galich A, et al.: Aberrant
J Biol Chem 273, 6166–6170, 1998. expression of E-cadherin and beta-catenin in human prostate cancer. Urol
8. Christofori G and Semb H: The role of the cell-adhesion molecule E- Oncol 23, 402–406, 2005.
cadherin as a tumour-suppressor gene. Trends Biochem Sci 24, 73–76, 27. Morita N, Uemura H, Tsumatani K, Cho M, Hirao Y, et al.: E-cadherin and
1999. alpha-, beta-, and gamma-catenin expression in prostate cancers: correlation
9. Paul R, Ewing CM, Jarrard DF, and Isaacs WB: The cadherin cell–cell with tumour invasion. Br J Cancer 79, 1879–1883, 1999.
adhesion pathway in prostate cancer progression. Br J Urol 79(Suppl 1), 28. Patriarca C, Petrella D, Campo B, Colombo P, Giunta P, et al.: Ele-
Downloaded by [University of Arizona] at 04:49 22 January 2013

37–43, 1997. vated E-cadherin and alpha/beta-catenin expression after androgen depri-
10. Playford MP, Bicknell D, Bodmer WF, and Macaulay VM: Insulin-like vation therapy in prostate adenocarcinoma. Pathol Res Pract 199, 659–665,
growth factor 1 regulates the location, stability, and transcriptional activity 2003.
of beta-catenin. Proc Natl Acad Sci USA 97, 12103–12108, 2000. 29. Nam S, Kim D, Cheng JQ, Zhang S, Lee JH, et al.: Action of the Src family
11. MacDonald BT, Tamai K, and He X: Wnt/beta-catenin signaling: compo- kinase inhibitor, dasatinib (BMS-354825), on human prostate cancer cells.
nents, mechanisms, and diseases. Dev Cell 17, 9–26, 2009. Cancer Res 65, 9185–9199, 2005.
12. Aberle H, Bauer A, Stappert J, Kispert A, and Kemler R: Beta-catenin is 30. Lilien J, Balsamo J, Arregui C, and Xu G: Turn-off, drop-out: functional
a target for the ubiquitin-proteasome pathway. EMBO J 16, 3797–3804, state switching of cadherins. Dev Dyn 224, 18–29, 2002.
1997. 31. Jamora C and Fuchs E: Intercellular adhesion, signaling and the cytoskele-
13. Yost C, Torres M, Miller JR, Huang E, Kimelman D, et al.: The axis- ton. Nat Cell Biol 4, E101–E108, 2002.
inducing activity, stability, and subcellular distribution of beta-catenin is 32. Chen HC, Chu RY, Hsu PN, Hsu PI, Lu JY, et al.: Loss of E-cadherin
regulated in Xenopus embryos by glycogen synthase kinase 3. Genes Dev expression correlates with poor differentiation and invasion into adjacent
10, 1443–1454, 1996. organs in gastric adenocarcinomas. Cancer Lett 201, 97–106, 2003.
14. Pollak MN, Schernhammer ES, and Hankinson SE: Insulin-like growth 33. Chesire DR and Isaacs WB: Beta-catenin signaling in prostate cancer: an
factors and neoplasia. Nat Rev Cancer 4, 505–518, 2004. early perspective. Endocr Relat Cancer 10, 537–560, 2003.
15. Hellawell GO, Turner GD, Davies DR, Poulsom R, Brewster SF, et al.: 34. Umbas R, Schalken JA, Aalders TW, Carter BS, Karthaus HF, et al.: Ex-
Expression of the type 1 insulin-like growth factor receptor is upregulated pression of the cellular adhesion molecule E-cadherin is reduced or absent
in primary prostate cancer and commonly persists in metastatic disease. in high-grade prostate cancer. Cancer Res 52, 5104–5109, 1992.
Cancer Res 62, 2942–2950, 2002. 35. Verras M and Sun Z: Roles and regulation of Wnt signaling and beta-catenin
16. Krueckl SL, Sikes RA, Edlund NM, Bell RH, Hurtado-Coll A, et al.: in prostate cancer. Cancer Lett 237, 22–32, 2006.
Increased insulin-like growth factor I receptor expression and signaling 36. El Touny LH and Banerjee PP: Akt–GSK-3 pathway as a target in
are components of androgen-independent progression in a lineage-derived genistein-induced inhibition of TRAMP prostate cancer progression to-
prostate cancer progression model. Cancer Res 64, 8620–8629, 2004. ward a poorly differentiated phenotype. Carcinogenesis 28, 1710–1717,
17. Nickerson T, Chang F, Lorimer D, Smeekens SP, Sawyers CL, et al.: In 2007.
vivo progression of LAPC-9 and LNCaP prostate cancer models to androgen 37. Wang Y, Kreisberg JI, and Ghosh PM: Cross-talk between the androgen
independence is associated with increased expression of insulin-like growth receptor and the phosphatidylinositol 3-kinase/Akt pathway in prostate
factor I (IGF-I) and IGF-I receptor (IGF-IR). Cancer Res 61, 6276–6280, cancer. Curr Cancer Drug Targets 7, 591–604, 2007.
2001. 38. Adlercreutz H, Markkanen H, and Watanabe S: Plasma concentrations of
18. Perabo FG, Von Low EC, Ellinger J, von Rucker A, Muller SC, et al.: Soy phyto-oestrogens in Japanese men. Lancet 13, 1209–1210, 1993.
isoflavone genistein in prevention and treatment of prostate cancer. Prostat 39. Uehar M, Arai Y, Watanabe S, and Adlercreutz H: Comparison of plasma
Cancer Prostatic Dis 11, 6–12, 2008. and urinary phytoestrogens in Japanese and Finnish women by time-
19. Baserga R: The insulin-like growth factor I receptor: a key to tumor growth? resolved fluoroimmunoassay. Biofactors 12, 217–225, 2000.
Cancer Res 55, 249–252, 1995. 40. Limer JL, Parkes AT, and Speirs V: Differential response to phytoestrogens
20. Pandini G, Mineo R, Frasca F, Roberts CT, Jr., Marcelli M, et al.: Androgens in endocrine sensitive and resistant breast cancer cells in vitro. Int J Cancer
upregulate the insulin-like growth factor-I receptor in prostate cancer cells. 119, 515–521, 2006.
Cancer Res 65, 1849–1857, 2005. 41. Moore AB, Castro L, Yu L, Zheng X, Di X, et al.: Stimulatory and inhibitory
21. Lee LF, Guan J, Qiu Y, and Kung HJ: Neuropeptide-induced androgen effects of genistein on human uterine leiomyoma cell proliferation are
independence in prostate cancer cells: roles of nonreceptor tyrosine kinases influenced by the concentration. Hum Reprod 22, 2623–2631, 2007.

You might also like